5 research outputs found

    Muscular and tendon degeneration after achilles rupture: new insights into future repair strategies

    Get PDF
    Achilles tendon rupture is a frequent injury with an increasing incidence. After clinical surgical repair, aimed at suturing the tendon stumps back into their original position, the repaired Achilles tendon is often plastically deformed and mechanically less strong than the pre-injured tissue, with muscle fatty degeneration contributing to function loss. Despite clinical outcomes, pre-clinical research has mainly focused on tendon structural repair, with a lack of knowledge regarding injury progression from tendon to muscle and its consequences on muscle degenerative/regenerative processes and function. Here, we characterize the morphological changes in the tendon, the myotendinous junction and muscle belly in a mouse model of Achilles tendon complete rupture, finding cellular and fatty infiltration, fibrotic tissue accumulation, muscle stem cell decline and collagen fiber disorganization. We use novel imaging technologies to accurately relate structural alterations in tendon fibers to pathological changes, which further explain the loss of muscle mechanical function after tendon rupture. The treatment of tendon injuries remains a challenge for orthopedics. Thus, the main goal of this study is to bridge the gap between clinicians'' knowledge and research to address the underlying pathophysiology of ruptured Achilles tendon and its consequences in the gastrocnemius. Such studies are necessary if current practices in regenerative medicine for Achilles tendon ruptures are to be improved

    Periosteum-derived mesenchymal progenitor cells in engineered implants promote fracture healing in a critical-size defect rat model

    No full text
    An attractive alternative to bone autografts is the use of autologous mesenchymal progenitor cells (MSCs) in combination with biomaterials. We compared the therapeutic potential of different sources of mesenchymal stem cells in combination with biomaterials in a bone nonunion model. A critical‐size defect was created in Sprague–Dawley rats. Animals were divided into six groups, depending on the treatment to be applied: bone defect was left empty (CTL); treated with live bone allograft (LBA); hrBMP‐2 in collagen scaffold (CSBMP2); acellular polycaprolactone scaffold (PCL group); PCL scaffold containing periosteum‐derived MSCs (PCLPMSCs) and PCL containing bone marrow‐derived MSCs (PCLBMSCs). To facilitate cell tracking, both MSCs and bone graft were isolated from green fluorescent protein (GFP)‐transgenic rats. CTL group did not show any signs of healing during the radiological follow‐up (n = 6). In the LBA group, all the animals showed bone bridging (n = 6) whereas in the CSBMP2 group, four out of six animals demonstrated healing. In PCL and PCLPMSCs groups, a reduced number of animals showed radiological healing, whereas no healing was detected in the PCLBMSCs group. Using microcomputed tomography, the bone volume filling the defect was quantified, showing significant new bone formation in the LBA, CSBMP2, and PCLPMSCs groups when compared with the CTL group. At 10 weeks, GFP positive cells were detected only in the LBA group and restricted to the outer cortical bone in close contact with the periosteum. Tracking of cellular implants demonstrated significant survival of the PMSCs when compared with BMSCs. In conclusion, PMSCs improve bone regeneration being suitable for mimetic autograft design

    Culture of human bone marrow-derived mesenchymal stem cells on of poly(L-lactic acid) scaffolds: potential application for the tissue engineering of cartilage

    Full text link
    Due to the attractive properties of poly(l-lactic acid) (PLLA) for tissue engineering, the aim was to determine the growth and differentiation capacity of mesenchymal stromal cells (MSCs) in PLLA scaffolds and their potential use in the treatment of cartilage diseases. MSCs were cultured in PLLA films and thin porous membranes to study adherence and proliferation. Permeability and porosity were determined for the different scaffolds employed. The optimal conditions for cell seeding were first determined, as well as cell density and distribution inside the PLLA. Scaffolds were then maintained in expansion or chondrogenic differentiation media for 21 days. Apoptosis, proliferation and chondrogenic differentiation was assessed after 21 days in culture by immunohistochemistry. Mechanical characteristics of scaffolds were determined before and after cell seeding. MSCs uniformly adhered to PLLA films as well as to porous membranes. Proliferation was detected only in monolayers of pure PLLA, but was no longer detected after 10 days. Mechanical characterization of PLLA scaffolds showed differences in the apparent compression elastic modulus for the two sizes used. After determining high efficiencies of seeding, the production of extracellular matrix (ECM) was determined and contained aggrecan and collagens type I and X. ECM produced by the cells induced a twofold increase in the apparent elastic modulus of the composite. Biocompatible PLLA scaffolds have been developed that can be efficiently loaded with MSCs. The scaffold supports chondrogenic differentiation and ECM deposition that improves the mechanics of the scaffold. Although this improvement does not met the expectations of a hyaline-like cartilage ECM, in part due to the lack of a mechanical stimulation, their potential use in the treatment of cartilage pathologies encourages to improve the mechanical component.This work has been supported by the Spanish Ministry of Science and Innovation DPI2010-20399-C04-00 project and Instituto de Salud Carlos III RETIC RD06/0014.Izal, I.; Aranda, P.; Sanz Ramos, P.; Ripalda, P.; Mora, G.; Granero Molto, F.; Deplaine, H.... (2013). Culture of human bone marrow-derived mesenchymal stem cells on of poly(L-lactic acid) scaffolds: potential application for the tissue engineering of cartilage. Knee Surgery, Sports Traumatology, Arthroscopy. 21(8):1737-1750. https://doi.org/10.1007/s00167-012-2148-6S17371750218Alberich-Bayarri A, Moratal D, Ivirico JL, Rodriguez-Hernandez JC, Valles-Lluch A, Marti-Bonmati L, Estelles JM, Mano JF, Pradas MM, Ribelles JL, Salmeron-Sanchez M (2009) Microcomputed tomography and microfinite element modeling for evaluating polymer scaffolds architecture and their mechanical properties. J Biomed Mater Res B Appl Biomater 91:191–202Aranda P, Agirre X, Ballestar E, Andreu EJ, Roman-Gomez J, Prieto I, Martin-Subero JI, Cigudosa JC, Siebert R, Esteller M, Prosper F (2009) Epigenetic signatures associated with different levels of differentiation potential in human stem cells. PLoS One 4:e7809Bentley G, Biant LC, Vijayan S, Macmull S, Skinner JA, Carrington RW (2012) Minimum ten-year results of a prospective randomised study of autologous chondrocyte implantation versus mosaicplasty for symptomatic articular cartilage lesions of the knee. J Bone Joint Surg Br 94:504–509Benya PD, Shaffer JD (1982) Dedifferentiated chondrocytes reexpress the differentiated collagen phenotype when cultured in agarose gels. Cell 30:215–224Breinan HA, Minas T, Hsu HP, Nehrer S, Shortkroff S, Spector M (2001) Autologous chondrocyte implantation in a canine model: change in composition of reparative tissue with time. J Orthop Res 19:482–492Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L (1994) Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med 331:889–895Bryant SJ, Anseth KS (2002) Hydrogel properties influence ECM production by chondrocytes photoencapsulated in poly(ethylene glycol) hydrogels. J Biomed Mater Res 59:63–72Budyanto L, Goh YQ, Ooi CP (2009) Fabrication of porous poly(l-lactide) (PLLA) scaffolds for tissue engineering using liquid–liquid phase separation and freeze extraction. J Mater Sci Mater Med 20:105–111Campoccia D, Doherty P, Radice M, Brun P, Abatangelo G, Williams DF (1998) Semisynthetic resorbable materials from hyaluronan esterification. Biomaterials 19:2101–2127Chen Y, Cho MR, Mak AF, Li JS, Wang M, Sun S (2008) Morphology and adhesion of mesenchymal stem cells on PLLA, apatite and apatite/collagen surfaces. J Mater Sci Mater Med 19:2563–2567Dar A, Shachar M, Leor J, Cohen S (2002) Optimization of cardiac cell seeding and distribution in 3D porous alginate scaffolds. Biotechnol Bioeng 80:305–312Deng Y, Zhao K, Zhang XF, Hu P, Chen GQ (2002) Study on the three-dimensional proliferation of rabbit articular cartilage-derived chondrocytes on polyhydroxyalkanoate scaffolds. Biomaterials 23:4049–4056Duance VC (1983) Surface of articular cartilage: immunohistological studies. Cell Biochem Funct 1:143–144Eerola I, Salminen H, Lammi P, Lammi M, von der Mark K, Vuorio E, Säämänen AM (1998) Type X collagen, a natural component of mouse articular cartilage: association with growth, aging, and osteoarthritis. Arthritis Rheum 41:1287–1295Filardo G, Kon E, Di Martino A, Iacono F, Marcacci M (2011) Arthroscopic second-generation autologous chondrocyte implantation: a prospective 7-year follow-up study. Am J Sports Med 39:2153–2160Freed LE, Marquis JC, Nohria A, Emmanual J, Mikos AG, Langer R (1993) Neocartilage formation in vitro and in vivo using cells cultured on synthetic biodegradable polymers. J Biomed Mater Res 27:11–23Freed LE, Vunjak-Novakovic G, Biron RJ, Eagles DB, Lesnoy DC, Barlow SK, Langer R (1994) Biodegradable polymer scaffolds for tissue engineering. Nat Biotechnol 12:689–693Gong Y, He L, Li J, Zhou Q, Ma Z, Gao C, Shen J (2007) Hydrogel-filled polylactide porous scaffolds for cartilage tissue engineering. J Biomed Mater Res B Appl Biomater 82:192–204Guo JF, Jourdian GW, Maccallum DK (1989) Culture and growth characteristics of chondrocytes encapsulated in alginate beads. Connect Tissue Res 19:277–297Ho MH, Kuo PY, Hsieh HJ, Hsien TY, Hou LT, Lai JY, Wang DM (2004) Preparation of porous scaffolds by using freeze-extraction and freeze-gelation methods. Biomaterials 25:129–138Hollister SJ, Lin CY, Saito E, Lin CY, Schek RD, Taboas JM, Williams JM, Partee B, Flanagan CL, Diggs A, Wilke EN, Van Lenthe GH, Müller R, Wirtz T, Das S, Feinberg SE, Krebsbach PH (2005) Engineering craniofacial scaffolds. Orthod Craniofac Res 8:162–173Holtzer H, Abbott J, Lash J, Holtzer S (1960) The loss of phenotypic traits by differentiated cells in vitro, I. Dedifferentiation of cartilage cells. Proc Natl Acad Sci USA 46:1533–1542Hsu SH, Tsai CL, Tang CM (2002) Evaluation of cellular affinity and compatibility to biodegradable polyesters and type-II collagen-modified scaffolds using immortalized rat chondrocytes. Artif Organs 26:647–658Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP (1997) Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitro. J Cell Biochem 64:295–312Jeong CG, Hollister SJ (2010) Mechanical and biochemical assessments of three-dimensional poly(1,8-octanediol-co-citrate) scaffold pore shape and permeability effects on in vitro chondrogenesis using primary chondrocytes. Tissue Eng Part A 16:3759–3768Jeong CG, Zhang H, Hollister SJ (2011) Three-dimensional poly(1,8-octanediol-co-citrate) scaffold pore shape and permeability effects on sub-cutaneous in vivo chondrogenesis using primary chondrocytes. Acta Biomater 7:505–514Kemppainen JM, Hollister SJ (2010) Differential effects of designed scaffold permeability on chondrogenesis by chondrocytes and bone marrow stromal cells. Biomaterials 31:279–287Langer R, Vacanti JP (1993) Tissue engineering. Science 260:920–926Lebourg M, Suay Antón J, Gómez Ribelles J (2008) Porous membranes of PLLA–PCL blend for tissue engineering applications. Eur Polymer J 44:2207–2218Lee CR, Breinan HA, Nehrer S, Spector M (2000) Articular cartilage chondrocytes in type I and type II collagen-GAG matrices exhibit contractile behavior in vitro. Tissue Eng 6:555–565Lee DA, Bentley G, Archer CW (1993) The control of cell division in articular chondrocytes. Osteoarthritis Cartilage 1:137–146Lee DA, Reisler T, Bader DL (2003) Expansion of chondrocytes for tissue engineering in alginate beads enhances chondrocytic phenotype compared to conventional monolayer techniques. Acta Orthop Scand 74:6–15Lennon DP, Haynesworth SE, Arm DM, Baber MA, Caplan AI (2000) Dilution of human mesenchymal stem cells with dermal fibroblasts and the effects on in vitro and in vivo osteochondrogenesis. Dev Dyn 219:50–62Li WJ, Tuli R, Okafor C, Derfoul A, Danielson KG, Hall DJ, Tuan RS (2005) A three-dimensional nanofibrous scaffold for cartilage tissue engineering using human mesenchymal stem cells. Biomaterials 26:599–609Ma Z, Gao C, Gong Y, Shen J (2005) Cartilage tissue engineering PLLA scaffold with surface immobilized collagen and basic fibroblast growth factor. Biomaterials 26:1253–1259Mackay AM, Beck SC, Murphy JM, Barry FP, Chichester CO, Pittenger MF (1998) Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng 4:415–428Marijnissen WJ, Van Osch GJ, Aigner J, van der Veen SW, Hollander AP, Verwoerd-Verhoef HL, Verhaar JA (2002) Alginate as a chondrocyte-delivery substance in combination with a non-woven scaffold for cartilage tissue engineering. Biomaterials 23:1511–1517Mayne R, Vail MS, Mayne PM, Miller EJ (1976) Changes in type of collagen synthesized as clones of chick chondrocytes grow and eventually lose division capacity. Proc Natl Acad Sci USA 73:1674–1678Mow VC, Holmes MH, Lai WM (1984) Fluid transport and mechanical properties of articular cartilage: a review. J Biomech 17:377–394Ochoa I, Sanz-Herrera JA, Garcia-Aznar JM, Doblare M, Yunos DM, Boccaccini AR (2009) Permeability evaluation of 45S5 bioglass-based scaffolds for bone tissue engineering. J Biomech 42:257–260Parkkinen JJ, Lammi MJ, Helminen HJ, Tammi M (1992) Local stimulation of proteoglycan synthesis in articular cartilage explants by dynamic compression in vitro. J Orthop Res 10:610–620Pérez Olmedilla M, Lebourg M, Escobar Ivirico JL, Nebot I, Garcia Giralt N, Gallego Ferrer G, Soria JM, Gómez Ribelles JL (2011) In vitro 3D culture of human chondrocytes using modified ε-caprolactone scaffolds with varying hydrophilicity and porosity. J Biomater Appl. doi: 10.1177/0885328211404263Perez-Ilzarbe M, Diez-Campelo M, Aranda P, Tabera S, Lopez T, Del Cañizo C, Merino J, Moreno C, Andreu EJ, Prosper F, Perez-Simon JA (2009) Comparison of ex vivo expansion culture conditions of mesenchymal stem cells for human cell therapy. Transfusion 49:1901–1910Peterson L, Vasiliadis HS, Brittberg M, Lindahl A (2010) Autologous chondrocyte implantation: a long-term follow-up. Am J Sports Med 38:1117–1124Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR (1999) Multilineage potential of adult human mesenchymal stem cells. Science 284:143–147Puppi D, Chiellini F, Piras AM, Chiellini E (2010) Polymeric materials for bone and cartilage repair. Prog Polym Sci 35:403–440Rotter N, Bucheler M, Haisch A, Wollenberg B, Lang S (2007) Cartilage tissue engineering using resorbable scaffolds. J Tissue Eng Regen Med 1:411–416Sah RL, Kim YJ, Doong JY, Grodzinsky AJ, Plaas AH, Sandy JD (1989) Biosynthetic response of cartilage explants to dynamic compression. J Orthop Res 7:619–636Saini S, Wick TM (2003) Concentric cylinder bioreactor for production of tissue engineered cartilage: effect of seeding density and hydrodynamic loading on construct development. Biotechnol Prog 19:510–521Shastri VP, Martin I, Langer R (2000) Macroporous polymer foams by hydrocarbon templating. Proc Natl Acad Sci USA 97:1970–1975Sherwooda JK, Riley SL, Palazzolo R, Brown SC, Monkhouse DC, Coates M, Griffith LG, Landeen LK, Ratcliffe A (2002) A three-dimensional osteochondral composite scaffold or articular cartilage repair. Biomaterials 23:4739–4751Shieh AC, Athanasiou KA (2003) Principles of cell mechanics for cartilage tissue engineering. Ann Biomed Eng 31:1–11Stenhamre H, Nannmark U, Lindahl A, Gatenholm P, Brittberg MJ (2010) Influence of pore size on the redifferentiation potential of human articular chondrocytes in poly(urethane urea) scaffolds. Tissue Eng Regen Med 5:578–588Temenoff JS, Mikos AG (2000) Review: tissue engineering for regeneration of articular cartilage. Biomaterials 21:431–440Thorogood P, Bee J, Von Der Mark K (1986) Transient expression of collagen type II at epitheliomesenchymal interfaces during morphogenesis of the cartilaginous neurocranium. Dev Biol 116:497–509Tsai WB, Chen CH, Chen JF, Chang KY (2006) The effects of types of degradable polymers on porcine chondrocyte adhesion, proliferation and gene expression. J Mater Sci Mater Med 17:337–343Von Der Mark K, Gauss V, Von Der Mark H, Muller P (1977) Relationship between cell shape and type of collagen synthesised as chondrocytes lose their cartilage phenotype in culture. Nature 267:531–532Weir NA, Buchanan FJ, Orr JF, Dickson GR (2004) Degradation of poly-l-lactide. Part 1: in vitro and in vivo physiological temperature degradation. Proc Inst Mech Eng [H] 218:307–319Yamane S, Iwasaki N, Kasahara Y, Harada K, Majima T, Monde K, Nishimura SI, Minami A (2007) Effect of pore size on in vitro cartilage formation using chitosan-based hyaluronic acid hybrid polymer fibers. J Biomed Mater Res A 81:586–593Zeltinger J, Sherwood JK, Graham DA, Mueller R, Griffith LG (2001) Effect of pore size and void fraction on cellular adhesion, proliferation, and matrix deposition. Tissue Eng 7:557–57

    Morphologic comparison of cervical, thoracic, lumbar intervertebral discs of cynomolgus monkey (Macaca fascicularis)

    No full text
    The aim was to analyze the morphological differences of the intervertebral disc and endplates at different levels. Forty-five vertebral motion segments were obtained from the spine of nine 3 to 4-year-old cynomolgus monkeys (Macaca fascicularis). From every spine, five discs were sectioned (C5–C6, T3–T4, T9–T10, L2–L3, L4–L5). In all the groups, tissue samples were collected and sections were stained with Masson’s trichrome, Safranine-O and van Gieson’s connective tissue stain to analyze the intervertebral discs. Immunohistochemistry was performed, using specific antibodies to detect collagens I and II. The intervertebral disc height, the maximum nucleus pulposus height, the superior and inferior endplate heights were histomorphometrically measured and different indexes were calculated to compare the differences between specimens of the same animal and between discs of the same level, and finally the differences between groups of discs of different levels. There were no differences existing in annular fibers anchoring on the endplate between discs of different levels. A positive immune reaction for type I collagen was observed in the longitudinal ligaments and in the annular region adjacent to them. Collagen II immune reactivity was found in the annulus close to the nucleus pulposus, in the endplates and in the nucleus. There were no differences between discs of different levels in the collagen I and II localization. The height of the discs varied along the spine. The smallest value was measured in T3–T4, with a larger increase caudally than cranially. The highest value was measured in L2–L3. A cervical disc was 55% the height of a lumbar one. The endplate height increased along the length of the spine. The inferior EP was always higher than the superior. The study provides a detailed structural characterization of the intervertebral disc and may be useful for further investigations on the disc degeneration process
    corecore